15 results on '"Lauren B. Arendse"'
Search Results
2. Probing the Requirements for Dual Angiotensin-Converting Enzyme C-Domain Selective/Neprilysin Inhibition
- Author
-
Lauren B. Arendse, Gyles E. Cozier, Charles J. Eyermann, Gregory S. Basarab, Sylva L. Schwager, Kelly Chibale, K. Ravi Acharya, and Edward D. Sturrock
- Subjects
Binding Sites ,Pyridines ,Thiazepines ,Angiotensin-Converting Enzyme Inhibitors ,Peptidyl-Dipeptidase A ,Bradykinin ,Crystallography, X-Ray ,Kinetics ,Lisinopril ,Drug Discovery ,Humans ,Molecular Medicine ,Computer Simulation ,Neprilysin - Abstract
Selective inhibition of the angiotensin-converting enzyme C-domain (cACE) and neprilysin (NEP), leaving the ACE N-domain (nACE) free to degrade bradykinin and other peptides, has the potential to provide the potent antihypertensive and cardioprotective benefits observed for nonselective dual ACE/NEP inhibitors, such as omapatrilat, without the increased risk of adverse effects. We have synthesized three 1-carboxy-3-phenylpropyl dipeptide inhibitors with nanomolar potency based on the previously reported C-domain selective ACE inhibitor lisinopril-tryptophan (LisW) to probe the structural requirements for potent dual cACE/NEP inhibition. Here we report the synthesis, enzyme kinetic data, and high-resolution crystal structures of these inhibitors bound to nACE and cACE, providing valuable insight into the factors driving potency and selectivity. Overall, these results highlight the importance of the interplay between the S1′ and S2′ subsites for ACE domain selectivity, providing guidance for future chemistry efforts toward the development of dual cACE/NEP inhibitors.
- Published
- 2022
3. The anticancer human mTOR inhibitor sapanisertib potently inhibits multiple Plasmodium kinases and life cycle stages
- Author
-
Lauren B. Arendse, James M. Murithi, Tarrick Qahash, Charisse Flerida A. Pasaje, Luiz C. Godoy, Sumanta Dey, Liezl Gibhard, Sonja Ghidelli-Disse, Gerard Drewes, Marcus Bantscheff, Maria J. Lafuente-Monasterio, Stephen Fienberg, Lynn Wambua, Samuel Gachuhi, Dina Coertzen, Mariëtte van der Watt, Janette Reader, Ayesha S. Aswat, Erica Erlank, Nelius Venter, Nimisha Mittal, Madeline R. Luth, Sabine Ottilie, Elizabeth A. Winzeler, Lizette L. Koekemoer, Lyn-Marie Birkholtz, Jacquin C. Niles, Manuel Llinás, David A. Fidock, and Kelly Chibale
- Subjects
General Medicine - Abstract
Compounds acting on multiple targets are critical to combating antimalarial drug resistance. Here, we report that the human “mammalian target of rapamycin” (mTOR) inhibitor sapanisertib has potent prophylactic liver stage activity, in vitro and in vivo asexual blood stage (ABS) activity, and transmission-blocking activity against the protozoan parasite Plasmodium spp. Chemoproteomics studies revealed multiple potential Plasmodium kinase targets, and potent inhibition of Plasmodium phosphatidylinositol 4-kinase type III beta (PI4Kβ) and cyclic guanosine monophosphate–dependent protein kinase (PKG) was confirmed in vitro. Conditional knockdown of PI4Kβ in ABS cultures modulated parasite sensitivity to sapanisertib, and laboratory-generated P. falciparum sapanisertib resistance was mediated by mutations in PI4Kβ. Parasite metabolomic perturbation profiles associated with sapanisertib and other known PI4Kβ and/or PKG inhibitors revealed similarities and differences between chemotypes, potentially caused by sapanisertib targeting multiple parasite kinases. The multistage activity of sapanisertib and its in vivo antimalarial efficacy, coupled with potent inhibition of at least two promising drug targets, provides an opportunity to reposition this pyrazolopyrimidine for malaria.
- Published
- 2022
4. The anticancer human mTOR inhibitor sapanisertib potently inhibits multiple
- Author
-
Lauren B, Arendse, James M, Murithi, Tarrick, Qahash, Charisse Flerida A, Pasaje, Luiz C, Godoy, Sumanta, Dey, Liezl, Gibhard, Sonja, Ghidelli-Disse, Gerard, Drewes, Marcus, Bantscheff, Maria J, Lafuente-Monasterio, Stephen, Fienberg, Lynn, Wambua, Samuel, Gachuhi, Dina, Coertzen, Mariëtte, van der Watt, Janette, Reader, Ayesha S, Aswat, Erica, Erlank, Nelius, Venter, Nimisha, Mittal, Madeline R, Luth, Sabine, Ottilie, Elizabeth A, Winzeler, Lizette L, Koekemoer, Lyn-Marie, Birkholtz, Jacquin C, Niles, Manuel, Llinás, David A, Fidock, and Kelly, Chibale
- Subjects
Sirolimus ,Mammals ,Antimalarials ,Life Cycle Stages ,Plasmodium ,TOR Serine-Threonine Kinases ,Plasmodium falciparum ,Guanosine Monophosphate ,Animals ,Humans ,MTOR Inhibitors ,1-Phosphatidylinositol 4-Kinase - Abstract
Compounds acting on multiple targets are critical to combating antimalarial drug resistance. Here, we report that the human "mammalian target of rapamycin" (mTOR) inhibitor sapanisertib has potent prophylactic liver stage activity, in vitro and in vivo asexual blood stage (ABS) activity, and transmission-blocking activity against the protozoan parasite
- Published
- 2022
5. Plasmodium Kinases as Potential Drug Targets for Malaria: Challenges and Opportunities
- Author
-
Kelly Chibale, Lauren B. Arendse, Ian H. Gilbert, and Susan Wyllie
- Subjects
0301 basic medicine ,Drug ,Plasmodium ,media_common.quotation_subject ,030106 microbiology ,Druggability ,malaria ,Computational biology ,lipid kinase ,drug discovery ,03 medical and health sciences ,parasitic diseases ,medicine ,Kinome ,Protein kinase A ,media_common ,biology ,Drug discovery ,Kinase ,protein kinase ,medicine.disease ,biology.organism_classification ,3. Good health ,030104 developmental biology ,Infectious Diseases ,target validation ,Perspective ,Malaria - Abstract
Protein and phosphoinositide kinases have been successfully exploited as drug targets in various disease areas, principally in oncology. In malaria, several protein kinases are under investigation as potential drug targets, and an inhibitor of Plasmodium phosphatidylinositol 4-kinase type III beta (PI4KIIIβ) is currently in phase 2 clinical studies. In this Perspective, we review the potential of kinases as drug targets for the treatment of malaria. Kinases are known to be readily druggable, and many are essential for parasite survival. A key challenge in the design of Plasmodium kinase inhibitors is obtaining selectivity over the corresponding human orthologue(s) and other human kinases due to the highly conserved nature of the shared ATP binding site. Notwithstanding this, there are some notable differences between the Plasmodium and human kinome that may be exploitable. There is also the potential for designed polypharmacology, where several Plasmodium kinases are inhibited by the same drug. Prior to starting the drug discovery process, it is important to carefully assess potential kinase targets to ensure that the inhibition of the desired kinase will kill the parasites in the required life-cycle stages with a sufficiently fast rate of kill. Here, we highlight key target attributes and experimental approaches to consider and summarize the progress that has been made targeting Plasmodium PI4KIIIβ, cGMP-dependent protein kinase, and cyclin-dependent-like kinase 3.
- Published
- 2021
6. New Amidated 3,6-Diphenylated Imidazopyridazines with Potent Antiplasmodium Activity Are Dual Inhibitors of Plasmodium Phosphatidylinositol-4-kinase and cGMP-Dependent Protein Kinase
- Author
-
Sergio Wittlin, Lynn Wambua, Luyanda Centani, Lauren B. Arendse, Lyn-Marie Birkholtz, Dale Taylor, Kelly Chibale, Janette Reader, Dina Coertzen, Nina Lawrence, Malkeet Kumar, Shoneeze S Renga, Peter Mubanga Cheuka, Stephen Fienberg, Godwin Akpeko Dziwornu, and Mariëtte van der Watt
- Subjects
0301 basic medicine ,biology ,Chemistry ,Kinase ,030106 microbiology ,hERG ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,Infectious Diseases ,Biochemistry ,In vivo ,biology.protein ,Phosphatidylinositol ,Protein kinase A ,cGMP-dependent protein kinase ,Adenosine triphosphate ,IC50 - Abstract
Recent studies on 3,6-diphenylated imidazopyridazines have demonstrated impressive in vitro activity and in vivo efficacy in mouse models of malaria infection. Herein, we report the synthesis and antiplasmodium evaluation of a new series of amidated analogues and demonstrate that these compounds potently inhibit Plasmodium phosphatidylinositol-4-kinase (PI4K) type IIIβ while moderately inhibiting cyclic guanidine monophosphate (cGMP)-dependent protein kinase (PKG) activity in vitro. Using in silico docking, we predict key binding interactions for these analogues within the adenosine triphosphate (ATP)-binding site of PI4K and PKG, paving the way for structure-based optimization of imidazopyridazines targeting both Plasmodium PI4K and PKG. While several derivatives showed low nanomolar antiplasmodium activity (IC50 < 100 nM), some compounds, including piperazine analogue 28, resulted in strong dual PI4K and PKG inhibition. The compounds also demonstrated transmission-blocking potential, evident from their potent inhibition of early- and late-stage gametocytes. Finally, the current compounds generally showed improved aqueous solubility and reduced hERG (human ether-a-go-go-related gene) channel inhibition.
- Published
- 2020
7. Structural Basis for Inhibitor Potency and Selectivity of Plasmodium falciparum Phosphatidylinositol 4-Kinase Inhibitors
- Author
-
John E. Burke, Jacob A. McPhail, Charles J. Eyermann, Kelly Chibale, Stephen Fienberg, Gregory S. Basarab, and Lauren B. Arendse
- Subjects
0301 basic medicine ,Drug ,biology ,Chemistry ,Kinase ,media_common.quotation_subject ,030106 microbiology ,Plasmodium falciparum ,Computational biology ,biology.organism_classification ,3. Good health ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,Infectious Diseases ,Docking (molecular) ,Potency ,Phosphatidylinositol ,Homology modeling ,Kinase activity ,media_common - Abstract
Plasmodium falciparum phosphatidylinositol 4-kinase (PfPI4K) has emerged as a promising new drug target for novel antimalarial therapeutics. In the absence of a reliable high-resolution three-dimensional structure, a homology model of PfPI4K was built as a tool for structure-based drug design. This homology model has been validated against three distinct chemical series of potent inhibitors using docking and energy minimizations to elucidate the interactions crucial for PI4K inhibition and potent antiplasmodium activity. Despite its potential as an antimalarial target, the similarity between PfPI4K and structurally related human kinases poses a risk for human off-target kinase activity and associated toxicity. Comparative docking between PfPI4K and human phosphoinositide kinases (PIKs) presents compelling evidence for the origins of selectivity. This in-depth analysis of the PfPI4K homology model, the binding modes of the inhibitors, and the interactions responsible for selectivity over human kinases provides a powerful template for future optimization of Plasmodium PI4K inhibitors.
- Published
- 2020
8. Selective inhibition of the C-domain of ACE (angiotensin-converting enzyme) combined with inhibition of NEP (neprilysin): a potential new therapy for hypertension
- Author
-
Augusto C. Montezano, Tomasz J. Guzik, Karla B Neves, Marko Poglitsch, Francisco J. Rios, Lauren B. Arendse, Rhian M. Touyz, Delyth Graham, Rheure Alves-Lopes, Dominik Skiba, Adam Harvey, and Edward D. Sturrock
- Subjects
0301 basic medicine ,Pyridines ,Thiazepines ,medicine.drug_class ,Antihypertensive Treatment ,Angiotensin-Converting Enzyme Inhibitors ,Blood Pressure ,Mice, Transgenic ,Vascular permeability ,030204 cardiovascular system & hematology ,Pharmacology ,Sacubitril ,neprilysin ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Lisinopril ,Renin ,Internal Medicine ,medicine ,Animals ,Antihypertensive drug ,Antihypertensive Agents ,omapatrilat ,business.industry ,Aminobutyrates ,Biphenyl Compounds ,Body Weight ,Original Articles ,Angiotensin II ,vasodilatation ,030104 developmental biology ,Blood pressure ,Liver ,Hypertension ,ACE inhibitor ,ComputingMethodologies_DOCUMENTANDTEXTPROCESSING ,Omapatrilat ,permeability ,business ,medicine.drug - Abstract
Supplemental Digital Content is available in the text., Combined inhibition of NEP (neutral endopeptidase) and ACE (angiotensin-converting enzyme), without unwanted effects, remains an attractive therapeutic strategy in cardiovascular medicine. Omapatrilat, a dual NEP inhibitor–ACE inhibitor, was a promising antihypertensive drug but failed in trials due to angioedema, an effect possibly caused by inhibition of both the N- and C-domains of ACE. Here, we aimed to determine whether lisinopril-tryptophan (lisW-S), a C-domain specific ACE inhibitor that preserves the N-domain catalytic activity, together with sacubitril (NEP inhibitor), differentially influences cardiovascular function and vascular permeability in hypertension compared with omapatrilat and lisinopril+sacubitril which inhibits both the ACE C- and N-domains. Ang II (angiotensin II)–dependent hypertensive mice (transgenic mice expressing active human renin in the liver [also known as LinA3]) received vehicle, sacubitril, lisW-S, lisinopril, lisinopril+sacubitril, or lisW-S+sacubitril for 4 weeks. Systolic blood pressure was increased in LinA3 mice, along with cardiac hypertrophy/dysfunction, impaired endothelium-dependent vasorelaxation, hypercontractile responses, vascular remodeling, and renal inflammation. LisW-S+sacubitril, lisinopril+sacubitril, and omapatrilat reduced systolic blood pressure and normalized cardiovascular remodeling and vascular hypercontractile responses in LinA3 mice. Although lisinopril+sacubitril and omapatrilat improved Ach-induced vasorelaxation, lisW-S+sacubitril had no effect. Endothelial permeability (Evans Blue assessment) was increased in omapatrilat but not in LisW-S+sacubitril–treated mice. In conclusion, lisW-S combined with sacubitril reduced systolic blood pressure and improved cardiac dysfunction in LinA3 mice, similar to omapatrilat but without effects on endothelium-dependent vasorelaxation. Moreover, increased vascular leakage (plasma extravasation) induced by omapatrilat was not evident in mice treated with lisW-S+sacubitril. Targeting ACE C-domain and NEP as a combination therapy may be as effective as omapatrilat in lowering systolic blood pressure, but without inducing vascular permeability and endothelial injury.
- Published
- 2021
9. New Amidated 3,6-Diphenylated Imidazopyridazines with Potent Antiplasmodium Activity Are Dual Inhibitors of
- Author
-
Peter Mubanga, Cheuka, Luyanda, Centani, Lauren B, Arendse, Stephen, Fienberg, Lynn, Wambua, Shoneeze S, Renga, Godwin Akpeko, Dziwornu, Malkeet, Kumar, Nina, Lawrence, Dale, Taylor, Sergio, Wittlin, Dina, Coertzen, Janette, Reader, Mariette, van der Watt, Lyn-Marie, Birkholtz, and Kelly, Chibale
- Subjects
Plasmodium ,Plasmodium falciparum ,Cyclic GMP-Dependent Protein Kinases ,Phosphatidylinositols ,1-Phosphatidylinositol 4-Kinase ,Protein Kinases ,Guanidine - Abstract
Recent studies on 3,6-diphenylated imidazopyridazines have demonstrated impressive
- Published
- 2020
10. Inhibition of resistance-refractory P. falciparum kinase PKG delivers prophylactic, blood stage, and transmission-blocking antiplasmodial activity
- Author
-
Jacquin C. Niles, Olalla Sanz, Anne-Catrin Uhlemann, Giulia Siciliano, Marcus C. S. Lee, Pietro Alano, Tomas Yeo, Michael J. Delves, Charisse Flerida A. Pasaje, Manuel Llinás, Sabine Ottilie, Louis Dwomoh, Kathryn J. Wicht, David A. Fidock, Megan J. Bird, Marla J. Giddins, Elizabeth A. Winzeler, Lauren B. Arendse, Nimisha Mittal, Emma F. Carpenter, T. R. Santha Kumar, Sonja Ghidelli-Disse, Natasha Spottiswoode, Sachel Mok, Edward Owen, Manu Vanaerschot, Kelly Chibale, James M. Murithi, Christian Doerig, Markus Bösche, and Andrew B. Tobin
- Subjects
Proteomics ,Clinical Biochemistry ,Drug Resistance ,Protozoan Proteins ,Pharmacology ,01 natural sciences ,Biochemistry ,law.invention ,Mice ,malaria drug discovery ,law ,Drug Discovery ,Tyrosine ,Mice, Inbred BALB C ,Gene knockdown ,Kinase ,Imidazoles ,Phosphoproteomics ,phosphoproteomics ,Molecular Docking Simulation ,Recombinant DNA ,cardiovascular system ,Molecular Medicine ,Female ,cGMP-dependent protein kinase (PKG) ,kinase ,Plasmodium falciparum ,Biology ,Article ,resistance ,Antimalarials ,Mediator ,target identification ,parasitic diseases ,Cyclic GMP-Dependent Protein Kinases ,Animals ,Humans ,Metabolomics ,Protein kinase A ,Molecular Biology ,Life Cycle Stages ,Binding Sites ,conditional knockdown ,010405 organic chemistry ,biology.organism_classification ,chemoproteomics ,0104 chemical sciences ,Hepatocytes - Abstract
Summary The search for antimalarial chemotypes with modes of action unrelated to existing drugs has intensified with the recent failure of first-line therapies across Southeast Asia. Here, we show that the trisubstituted imidazole MMV030084 potently inhibits hepatocyte invasion by Plasmodium sporozoites, merozoite egress from asexual blood stage schizonts, and male gamete exflagellation. Metabolomic, phosphoproteomic, and chemoproteomic studies, validated with conditional knockdown parasites, molecular docking, and recombinant kinase assays, identified cGMP-dependent protein kinase (PKG) as the primary target of MMV030084. PKG is known to play essential roles in Plasmodium invasion of and egress from host cells, matching MMV030084's activity profile. Resistance selections and gene editing identified tyrosine kinase-like protein 3 as a low-level resistance mediator for PKG inhibitors, while PKG itself never mutated under pressure. These studies highlight PKG as a resistance-refractory antimalarial target throughout the Plasmodium life cycle and promote MMV030084 as a promising Plasmodium PKG-targeting chemotype., Graphical Abstract, Highlights • MMV030084 inhibits P. falciparum liver and asexual blood stages and male gametes • Proteomic and conditional knockdown studies identified PfPKG as the target • Resistance selection studies identified TKL3 as a low-level resistance mediator • PKG is a promising resistance-refractory target for antimalarial drug development, Vanaerschot et al. report an antimalarial, MMV030084, with potent antiplasmodial activity against all stages of human infection by Plasmodium falciparum. Metabolomic, phosphoproteomic, chemoproteomic, and gene-editing studies identified cGMP-dependent protein kinase (PKG) as the primary target, which did not mutate under selective drug pressure.
- Published
- 2020
11. Effects of polymorphic variation on the thermostability of heterogenous populations of CYP3A4 and CYP2C9 enzymes in solution
- Author
-
Jonathan M. Blackburn and Lauren B. Arendse
- Subjects
0301 basic medicine ,In silico ,lcsh:Medicine ,Single-nucleotide polymorphism ,Oligomer ,Polymorphism, Single Nucleotide ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Cytochrome P-450 CYP3A ,Humans ,Destabilisation ,lcsh:Science ,Cells, Cultured ,Thermostability ,Cytochrome P-450 CYP2C9 ,chemistry.chemical_classification ,Multidisciplinary ,biology ,CYP3A4 ,Chemistry ,Protein Stability ,lcsh:R ,Cytochrome P450 ,Kinetics ,030104 developmental biology ,Enzyme ,Biochemistry ,biology.protein ,lcsh:Q - Abstract
The effect of non-synonymous single nucleotide polymorphisms (SNPs) on cytochrome P450 (CYP450) drug metabolism is currently poorly understood due to the large number of polymorphisms, the diversity of potential substrates and the complexity of CYP450 function. Previously we carried out in silico studies to explore the effect of SNPs on CYP450 function, using in silico calculations to predict the effect of mutations on protein stability. Here we have determined the effect of eight CYP3A4 and seven CYP2C9 SNPs on the thermostability of proteins in solution to test these predictions. Thermostability assays revealed distinct CYP450 sub-populations with only 65–70% of wild-type CYP3A4 and CYP2C9 susceptible to rapid heat-induced P450 to P420 conversion. CYP3A4 mutations G56D, P218R, S222P, I223R, L373F and M445T and CYP2C9 mutations V76M, I359L and I359T were destabilising, increasing the proportion of protein sensitive to the rapid heat-induced P450 to P420 conversion and/or reducing the half-life of this conversion. CYP2C9 Q214L was the only stabilising mutation. These results corresponded well with the in silico protein stability calculations, confirming the value of these predictions and together suggest that the changes in thermostability result from destabilisation/stabilisation of the protein fold, changes in the haem-binding environment or effects on oligomer formation/conformation.
- Published
- 2018
12. Structural basis for the C-domain-selective angiotensin-converting enzyme inhibition by bradykinin-potentiating peptide b (BPPb)
- Author
-
Lauren B. Arendse, Lizelle Lubbe, Sylva L. U. Schwager, Afolake T. Arowolo, Emma Ruth Belcher, Gyles E. Cozier, Edward D. Sturrock, and K. Ravi Acharya
- Subjects
Bradykinin ,Peptide ,Angiotensin-Converting Enzyme Inhibitors ,CHO Cells ,Peptidyl-Dipeptidase A ,Crystallography, X-Ray ,Biochemistry ,Catalysis ,03 medical and health sciences ,chemistry.chemical_compound ,Cricetulus ,Protein Domains ,Hydrolase ,Animals ,Humans ,Enzyme kinetics ,Molecular Biology ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Metalloproteinase ,biology ,030302 biochemistry & molecular biology ,Active site ,Angiotensin-converting enzyme ,Cell Biology ,Enzyme ,chemistry ,biology.protein ,Mutagenesis, Site-Directed ,Oligopeptides - Abstract
Angiotensin-converting enzyme (ACE) is a zinc metalloprotease best known for its role in blood pressure regulation. ACE consists of two homologous catalytic domains, the N- and C-domain, that display distinct but overlapping catalytic functions in vivo owing to subtle differences in substrate specificity. While current generation ACE inhibitors target both ACE domains, domain-selective ACE inhibitors may be clinically advantageous, either reducing side effects or having utility in new indications. Here, we used site-directed mutagenesis, an ACE chimera and X-ray crystallography to unveil the molecular basis for C-domain-selective ACE inhibition by the bradykinin-potentiating peptide b (BPPb), naturally present in Brazilian pit viper venom. We present the BPPb N-domain structure in comparison with the previously reported BPPb C-domain structure and highlight key differences in peptide interactions with the S4 to S9 subsites. This suggests the involvement of these subsites in conferring C-domain-selective BPPb binding, in agreement with the mutagenesis results where unique residues governing differences in active site exposure, lid structure and dynamics between the two domains were the major drivers for C-domain-selective BPPb binding. Mere disruption of BPPb interactions with unique S2 and S4 subsite residues, which synergistically assist in BPPb binding, was insufficient to abolish C-domain selectivity. The combination of unique S9–S4 and S2′ subsite C-domain residues was required for the favourable entry, orientation and thus, selective binding of the peptide. This emphasizes the need to consider factors other than direct protein–inhibitor interactions to guide the design of domain-selective ACE inhibitors, especially in the case of larger peptides.
- Published
- 2019
13. INHIBITION OF ACE C-DOMAIN AND NEPRILYSIN AS A NEW THERAPY IN HYPERTENSION
- Author
-
Lauren B. Arendse, Francisco J. Rios, Edward D. Sturrock, Rhian M. Touyz, Mario R. W. Ehlers, Augusto C. Montezano, Livia L Camargo, Karla B Neves, Marko Poglitsch, Delyth Graham, Rheure Alves-Lopes, and Adam Harvey
- Subjects
Physiology ,business.industry ,Internal Medicine ,Medicine ,Computational biology ,Cardiology and Cardiovascular Medicine ,business ,Neprilysin ,Domain (software engineering) - Published
- 2021
14. Molecular Basis for Multiple Omapatrilat Binding Sites within the ACE C-Domain: Implications for Drug Design
- Author
-
Gyles E, Cozier, Lauren B, Arendse, Sylva L, Schwager, Edward D, Sturrock, and K Ravi, Acharya
- Subjects
Models, Molecular ,Pyridines ,Thiazepines ,Catalytic Domain ,Drug Design ,Humans ,Amino Acid Sequence ,Peptidyl-Dipeptidase A ,Ligands - Abstract
Omapatrilat was designed as a vasopeptidase inhibitor with dual activity against the zinc metallopeptidases angiotensin-1 converting enzyme (ACE) and neprilysin (NEP). ACE has two homologous catalytic domains (nACE and cACE), which exhibit different substrate specificities. Here, we report high-resolution crystal structures of omapatrilat in complex with nACE and cACE and show omapatrilat has subnanomolar affinity for both domains. The structures show nearly identical binding interactions for omapatrilat in each domain, explaining the lack of domain selectivity. The cACE complex structure revealed an omapatrilat dimer occupying the cavity beyond the S
- Published
- 2018
15. Combining in silico protein stability calculations with structure-function relationships to explore the effect of polymorphic variation on cytochrome P450 drug metabolism
- Author
-
Jonathan M. Blackburn, Lauren B. Arendse, and Tom L. Blundell
- Subjects
Models, Molecular ,Protein Conformation ,In silico ,Clinical Biochemistry ,Biology ,medicine.disease_cause ,Polymorphism, Single Nucleotide ,Structure-Activity Relationship ,Protein structure ,Cytochrome P-450 Enzyme System ,medicine ,Animals ,Humans ,Computer Simulation ,NADPH-Ferrihemoprotein Reductase ,Pharmacology ,Genetics ,Mutation ,Protein Stability ,Mutagenesis ,Cytochrome P450 reductase ,Cytochrome P450 ,Enzyme structure ,Rats ,Pharmaceutical Preparations ,biology.protein ,Mutagenesis, Site-Directed ,Drug metabolism - Abstract
We carried out an in silico structural analysis of 348 non-synonymous single nucleotide polymorphisms, found across nine of the major human drug metabolising cytochrome P450 isoforms, to determine the effects of mutations on enzyme structure and function. Previous functional studies in our group have delineated regions of the cytochrome P450 structure important for substrate recognition, substrate and product access and egress from the active site and interaction with the cytochrome P450 reductase. Here we combine the information from those studies with new in silico calculations on the effect of mutations on protein stability and we compare our results to experimental data in order to establish the likely causes of altered drug metabolism observed for cytochrome P450 variants in functional assays to date, in the process creating a cytochrome P450 polymorphic variant map. Using the computational tool Site Directed Mutator we predicted destabilising mutations that result in altered enzyme function in vitro with a specificity of 83%. We found that 75% of all cytochrome P450 mutations that show altered activity in vitro are either predicted to be destabilising to protein structure or are found within regions predicted to be important for catalytic activity. Furthermore, we found that 70% of the mutations that showed similar activity to the wild-type enzyme in in vitro studies lie outside of functional regions important for catalytic activity and are predicted to have no effect on protein stability. Our resultant cytochrome P450 polymorphic variant map should therefore find utility in predicting the likely functional effect of uncharacterised variants on drug metabolism.
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.